adoptive t cell therapy
Recently Published Documents


TOTAL DOCUMENTS

482
(FIVE YEARS 164)

H-INDEX

46
(FIVE YEARS 9)

Author(s):  
Christopher Aled Chamberlain ◽  
Eric Paul Bennett ◽  
Anders Handrup Kverneland ◽  
Inge Marie Svane ◽  
Marco Donia ◽  
...  

2021 ◽  
Author(s):  
Qian Zhu ◽  
guoliang qiao ◽  
Lefu Huang ◽  
Chang Xu ◽  
Deliang Guo ◽  
...  

Abstract BackgroundsWe aimed at to investigate that restoring the amount of CD8+PD1+ T cells through adoptive T cell therapy (ACT) could improve the prognosis and facilitate the therapeutic response to anti-PD-1 in patients with advanced pancreatic cancer (APC).Methods177 adult patients who underwent tumor resection as initial treatment for PDAC during February 2013 to July 2019. at Zhongnan Hospital of Wuhan University were enrolled in this study. Another cohort of 32 patients with APC were prospectively enrolled from Capital Medical University Cancer Center, Beijing Shijitan Hospital from June 1, 2013, to May 30, 2019. All patients with APC underwent ACT and 15 of 32 (46.8%) patients received ACT combined with anti-PD-1 (Pembrolizumab).ResultsOf the 177 patients received tumor resection, 67 tumor samples showed overexpression of PD-L1 We found that high PD-L1 expression in tumor tissues was significantly associated with short overall survival. Also, we tested the percentage of peripheral CD8+PD-1+ T cells and found it was significantly correlated with the PD-L1 expression and the prognosis of patients with PDAC. We further tracked the peripheral blood T lymphocyte subtypes for 30 months and found that CD8+PD-1+ cells were decreased and we hypothesized that the CD8+PD-1+ cells were exhausted. After that, we performed ACT for patients with APC and we found that the ratios of post treatment of ACT/pre-ACT CD8+PD-1+ T cells were significantly related with the prognosis of patients with APC. Moreover, patients with combined treatment of ACT with anti-PD-1 had significantly favorable both OS and PFS. Furthermore, T-cell receptor (TCR) repertoire were tested and TCR diversity of cultured T cells were calculated and we found the treatment of ACT impacted on the TCR repertoire especially in patients with significantly CD8+PD-1+ T cells enhanced.Conclusionsthis study showed that the CD8+PD-1+ T cell subgroup was related with expression of PD-L1 and the prognosis of patients with PDAC who received surgical resection. The CD8+PD-1+ T cells were gradually exhausted and restoring it by treatment of ACT was associated with a significantly favorable prognosis and facilitate the response to Anti-PD-1.


2021 ◽  
Vol 22 (22) ◽  
pp. 12295
Author(s):  
Lewis Zhichang Shi ◽  
James A. Bonner

The unprecedented successes of immunotherapies (IOs) including immune checkpoint blockers (ICBs) and adoptive T-cell therapy (ACT) in patients with late-stage cancer provide proof-of-principle evidence that harnessing the immune system, in particular T cells, can be an effective approach to eradicate cancer. This instills strong interests in understanding the immunomodulatory effects of radiotherapy (RT), an area that was actually investigated more than a century ago but had been largely ignored for many decades. With the “newly” discovered immunogenic responses from RT, numerous endeavors have been undertaken to combine RT with IOs, in order to bolster anti-tumor immunity. However, the underlying mechanisms are not well defined, which is a subject of much investigation. We therefore conducted a systematic literature search on the molecular underpinnings of RT-induced immunomodulation and IOs, which identified the IFN–JAK–STAT pathway as a major regulator. Our further analysis of relevant studies revealed that the signaling strength and duration of this pathway in response to RT and IOs may determine eventual immunological outcomes. We propose that strategic targeting of this axis can boost the immunostimulatory effects of RT and radiosensitizing effects of IOs, thereby promoting the efficacy of combination therapy of RT and IOs.


2021 ◽  
Author(s):  
Malcolm J. W. Sim ◽  
Zachary Stotz ◽  
Jinghua Lu ◽  
Paul Brennan ◽  
Eric O. Long ◽  
...  

Dimorphic residues at positions 77 and 80 delineate HLA-C allotypes into two groups, C1 and C2, which associate with disease through interactions with C1 and C2-specific natural killer cell receptors. How the C1/C2 dimorphism affects T cell recognition is unknown. Using HLA-C allotypes that differ only by the C1/C2-defining residues, we found that KRAS-G12D neoantigen specific T cell receptors (TCR) discriminated groups C1 and C2 HLA-C, due to effects on peptide presentation and TCR affinity. Structural and functional experiments combined with immunopeptidomics analysis revealed that C1-HLA-C favors smaller amino acids at the peptide C-terminus minus-1 position (pΩ-1), and that larger pΩ-1 residues diminished TCR recognition of C1-HLA-C. After controlling for peptide presentation, TCRs exhibited weaker affinities for C2-HLA-C despite conserved TCR contacts. Thus, the C1/C2 dimorphism impacts peptide presentation and HLA-C restricted T cell responses, with implications in multiple disease contexts including adoptive T cell therapy targeting KRAS-G12D-induced cancers.


Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 3070
Author(s):  
Emilie Biele ◽  
Sebastian J. Schober ◽  
Carolin Prexler ◽  
Melanie Thiede ◽  
Kristina von Heyking ◽  
...  

Ewing’s sarcoma (EwS) is a pediatric solid tumor entity with low somatic mutational burden and a low rate of tumor-infiltrating T cells, indicating a low extent of immunogenicity. In EwS, immunogenicity may furthermore be significantly diminished by a predominantly M2 macrophage driven pro-tumorigenic tumor microenvironment. In the past, we demonstrated that CHM1319-specific TCR-transgenic T cells are able to control EwS growth in a preclinical mouse model as well as in a patient with metastatic disease. However, new adjuvant techniques to induce long lasting and curative CHM1319-specific TCR-transgenic T cell-mediated anti-tumor responses are needed. In this work, we sought to identify a technique to improve the cytotoxic effect of CHM1319-specific TCR-transgenic T cell by altering the immunogenic cell surface marker expression on EwS cell lines using different cytokines. We demonstrate that TNF, IL-6, IL-1β and PGE2 cause pro-immunogenic CD83, MHC class I and II as well as ICAM-1 upregulation in EwS cell lines. This observation was associated with significantly improved recognition and killing of the tumor cells by EwS-specific CHM1319/HLA-A*02:01-restricted TCR-transgenic T cells. Conclusively, we demonstrate that the induction of an inflammatory signature renders EwS more susceptible to adoptive T cell therapy. TNF, which is upregulated during inflammatory processes, is of particular translational interest as its secretion may be induced in the patients e.g., by irradiation and hyperthermia in the clinical setting. In future clinical protocols, this finding may be important to identify appropriate conditioning regimens as well as point of time for adoptive T cell-based immunotherapy in EwS patients.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi93-vi93
Author(s):  
Akane Yamamichi ◽  
Polly Chuntova ◽  
Bunta Kakihara ◽  
Tiffany Chen ◽  
David Diebold ◽  
...  

Abstract We have identified a novel HLA-A*02:01-restricted CD8 T-cell epitope encompassing the H3.3K27M mutation and a corresponding high-affinity T-cell receptor (TCR) that recognizes the epitope. While the development of adoptive cell transfer therapy using TCR-transduced T-cells holds a promise, we still need to overcome multiple challenges, such as suboptimal T-cell trafficking and the immunosuppressive environment of malignant glioma. For example, tumor-associated macrophages (TAMs) mediate immunosuppression but do not function as effective antigen-presenting cells. We have developed a novel cholesteryl pullulan (CHP) nanogel as a highly biocompatible and efficient vaccine delivery system targeting TAMs. In this study, we investigated whether the CHP nanogel loaded with the H3.3K27M peptide would deliver the peptide to TAMs and convert TAMs to better antigen-presenting cells that enhance the anti- H3.3K27M+ glioma activity of the TCR-transduced T-cells. As a clinically relevant mouse model, we used HLA-A2/HLA-DR1-transgenic mice and generated a syngeneic glioma cell line that expresses H3.3K27M from their astrocytes. We also generated a retroviral vector encoding the H3.3K27M-specific TCR for transduction of mouse T cells. HLA-A2/HLA-DR1-transgenic mice bearing day 16 intracerebral H3.3K27M+ glioma received an intravenous administration of the CHP nanogel along with poly-ICLC, a Toll-like receptor 3 agonist. The mice then received an intravenous infusion of TCR-transduced or control, non-transduced T-cells on the following day. The triple combination regimen with the CHP, poly-ICLC and TCR-transduced T-cells significantly suppressed the tumor growth, associated with increased levels of T-cell infiltration into the tumors compared with the dual-therapy with poly-ICLC and TCR-T-cells without the CHP. Furthermore, TAMs isolated from CHP-treated mice showed evidence of CHP-uptake, abilities to stimulate proliferation of TCR-transduced T-cells, and higher levels of HLA.A2 expression. These results suggest that the antigen-loaded CHP nanogel can promote the local antigen-presentation to T-cells and represent a promising approach for improving the efficacy of adoptive T-cell therapy for gliomas.


2021 ◽  
Vol 21 ◽  
Author(s):  
Ruby Dwivedi ◽  
Divya Mehrotra ◽  
Shaleen Chandra ◽  
Rahul Pandey

Backgound and Objective: Early chemoprevention in Oral Potentially Malignant Disorders (OPMDs) and Oral Cancer (OC) has been extensively researched in order to mitigate the malignant transformation and progression of the lesion. Many agents have been attempted, but their cost inefficacy and inadequate outcomes posed a major hindrance in their successful adoption. Retinoid Based Therapy (RBT) though a cheap and effective treatment option, could not achieve much clinical usage because of variable responsiveness in clinical outcomes. Such clinical response variability may be attributed to the repression of retinoid receptors by Preferentially Expressed Antigen of Melanoma (PRAME) protein molecule. Therefore, in order to make RBT successful, targeting PRAME by various immunotherapies is an exciting area of research investigation. This review provides an insight into the various immunotherapeutic strategies targeting PRAME and their usefulness in retinoid-resistant OPMD and OC. Method of data collection: An exhaustive internet-based literature search following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines was carried out in PUBMED and Google SCHOLAR database using terms ‘Anti-PRAME’ OR ‘PRAME Immunotherapy’ OR ‘PRAME Vaccines’ AND ‘Cancer’ AND ‘Retinoid resistance’. Only articles in the English language with at least 1 citation, published in a journal with impact factor ≥ 1, having relevance to the context and availability of full text were considered. Results: After an initial search, 342 articles were yielded on the basis of inclusion criteria and, by reading the abstract and availability of full text, a total of 124 articles were selected. Further reading the full texts and considering articles from the references of the selected articles, a total of 65 articles were finally included in the review. Conclusion: Our analysis of the literature suggests that PRAME screening in OC and OPMDs prior to RBT should be done. In PRAME positive cases, approaches like PRAME based immunotherapy in the form of Cancer vaccine therapy [Acellular PRAME vaccine, PRAME pulsed Dendritic Cells (DC)]; Adoptive T Cell therapy/T Cell Receptor-T Cell therapy, Antibody therapy/Chimeric Antigen Receptor-T Cell therapy along with Presented antigen modulation Therapies employing histone deacetylase inhibitors and demethylation agents seem plausible. In the future, a combination therapy employing either PRAME vaccines or antibodies or Adoptive T cell Therapy and ATRA could be used in retinoid resistant OC and OPMDs.


2021 ◽  
Vol 12 ◽  
Author(s):  
Danyang Wang ◽  
Hangyu Zhang ◽  
Tao Xiang ◽  
Gang Wang

Colorectal cancer (CRC) is one of the most common cancers worldwide. However, the treatment outcomes of immunotherapy in microsatellite-stable (MSS) CRC remain unsatisfactory. As the majority of CRC cases display a molecular MSS/mismatch repair-proficient (pMMR) profile, it is particularly meaningful to explore the clinical applications of adaptive immune therapy in MSS CRC patients. In this review, we summarized the therapeutic approaches of adoptive immune therapies, including cytokines, therapeutic cancer vaccines, adoptive T-cell therapy, and immune checkpoint inhibitors, in the treatment of MSS CRCs.


Sign in / Sign up

Export Citation Format

Share Document